Integrative Kinome Profiling Identifies Mtorc1/2 Inhibition As Treatment Strategy in Ovarian Clear Cell Carcinoma

Total Page:16

File Type:pdf, Size:1020Kb

Integrative Kinome Profiling Identifies Mtorc1/2 Inhibition As Treatment Strategy in Ovarian Clear Cell Carcinoma Published OnlineFirst April 23, 2018; DOI: 10.1158/1078-0432.CCR-17-3060 Cancer Therapy: Preclinical Clinical Cancer Research Integrative Kinome Profiling Identifies mTORC1/2 Inhibition as Treatment Strategy in Ovarian Clear Cell Carcinoma Joseph J. Caumanns1, Katrien Berns2, G. Bea A. Wisman1, Rudolf S.N. Fehrmann3, Tushar Tomar1, Harry Klip1, Gert J. Meersma1, E. Marielle Hijmans2, Annemiek M.C. Gennissen2, Evelien W. Duiker4, Desiree Weening5, Hiroaki Itamochi6, Roelof J.C. Kluin2, Anna K.L. Reyners3, Michael J. Birrer7, Helga B. Salvesen8, Ignace Vergote9, Els van Nieuwenhuysen9, James Brenton10, E. Ioana Braicu11, Jolanta Kupryjanczyk12, Beata Spiewankiewicz13, Lorenza Mittempergher2, Rene Bernards2, Ate G.J. van der Zee1, and Steven de Jong3 Abstract Purpose: Advanced-stage ovarian clear cell carcinoma Results: We identified several putative driver mutations in (OCCC) is unresponsive to conventional platinum-based che- kinases at low frequency that were not previously annotated in motherapy. Frequent alterations in OCCC include deleterious OCCC. Combining mutations and copy-number alterations, 91% mutations in the tumor suppressor ARID1A and activating of all tumors are affected in the PI3K/AKT/mTOR pathway, mutations in the PI3K subunit PIK3CA.Inthisstudy,weaimed the MAPK pathway, or the ERBB family of receptor tyrosine to identify currently unknown mutated kinases in patients with kinases, and 82% in the DNA repair pathway. Strong p-S6 staining OCCC and test druggability of downstream affected pathways in patients with OCCC suggests high mTORC1/2 activity. We in OCCC models. consistently found that the majority of OCCC cell lines are Experimental Design: In a large set of patients with OCCC (n ¼ especially sensitive to mTORC1/2 inhibition by AZD8055 and 124), the human kinome (518 kinases) and additional cancer- not toward drugs targeting ERBB family of receptor tyrosine related genes were sequenced, and copy-number alterations were kinases or DNA repair signaling. We subsequently demonstrated determined. Genetically characterized OCCC cell lines (n ¼ 17) the efficacy of mTORC1/2 inhibition in all our unique OCCC and OCCC patient–derived xenografts (n ¼ 3) were used for drug patient–derived xenograft models. testing of ERBB tyrosine kinase inhibitors erlotinib and lapatinib, Conclusions: These results propose mTORC1/2 inhibition as the PARP inhibitor olaparib, and the mTORC1/2 inhibitor an effective treatment strategy in OCCC. Clin Cancer Res; 1–13. Ó2018 AZD8055. AACR. Introduction ian cancer. The majority of patients with OCCC are diagnosed at In the United States, ovarian cancer is the fifth-leading cause of an early stage (57%–81% at stage I/II) and have better overall cancer deaths in women (1). Ovarian clear cell carcinoma survival compared with stage-matched high-grade serous (HGS) (OCCC) is the second most common subtype of epithelial ovar- ovarian cancer, the most common subtype of ovarian cancer. In 1Department of Gynecologic Oncology, Cancer Research Center Groningen, Medical University, Berlin, Germany. 12Department of Pathology and Laboratory University Medical Center Groningen, University of Groningen, Groningen, the Diagnostics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Netherlands. 2Division of Molecular Carcinogenesis, The Netherlands Cancer Oncology, Warsaw, Poland. 13Department of Gynecologic Oncology, Maria Institute, Amsterdam, the Netherlands. 3Department of Medical Oncology, Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Cancer Research Center Groningen, University Medical Center Groningen, Poland. University of Groningen, Groningen, the Netherlands. 4Department of Pathology and Medical Biology, Cancer Research Center Groningen, University Medical Note: Supplementary data for this article are available at Clinical Cancer Center Groningen, University of Groningen, Groningen, the Netherlands. Research Online (http://clincancerres.aacrjournals.org/). 5Department of Genetics, Cancer Research Center Groningen, University Med- ical Center Groningen, University of Groningen, Groningen, the Netherlands. In memory of H. Klip and H.B. Salvesen. 6Department of Obstetrics and Gynecology, Iwate Medical University School of Corresponding Author: Steven de Jong, Cancer Research Center Groningen, Medicine, Morioka, Iwate, Japan. 7Center for Cancer Research, The Gillette University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 Center for Gynecologic Oncology, Massachusetts General Hospital, Harvard GZ Groningen, the Netherlands. Phone: 31-50-3612964; Fax: 31-50-3614862; Medical School, Boston, Massachusetts. 8Department of Obstetrics and Gyne- E-mail: [email protected] cology, Haukeland University Hospital, Bergen, Norway. 9Department of Gynae- cology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven, doi: 10.1158/1078-0432.CCR-17-3060 Leuven, Belgium. 10Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom. 11Department of Gynecology, Charite Ó2018 American Association for Cancer Research. www.aacrjournals.org OF1 Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst April 23, 2018; DOI: 10.1158/1078-0432.CCR-17-3060 Caumanns et al. research in OCCC cell lines demonstrated sensitivity to PI3K/ Translational Relevance mTOR dual inhibitors and AKT inhibitors, although PIK3CA Advanced-stage ovarian clear cell carcinoma (OCCC) is less mutations did not predict sensitivity to these inhibitors (16, 17). responsive to platinum-based chemotherapy compared with In the present study, we aimed to identify novel targetable high-grade serous ovarian carcinoma. Our in-depth analyses mutations by means of high-coverage sequencing of all protein of a large set of patients with OCCC reveal numerous genomic kinase genes, referred to as the kinome, and of a subgroup of alterations related to activation of mTORC1/2. High sensitiv- cancer-related genes in a large set of OCCC. In addition, we ity, especially to inhibitors targeting both mTORC1 and determined copy-number gains and losses in kinases and other mTORC2, is observed in a large OCCC cell line panel. Similar genes of OCCC tumors using high-coverage single-nucleotide results are obtained in OCCC patient–derived xenografts, polymorphism (SNP) arrays. To detect kinase mutations and signifying the clinical implications of these genomic altera- CNA at both high and low frequencies, we used a large cohort tions. Targeting of mTORC1 in combination with standard of 124 untreated primary OCCC tumors and most of the available chemotherapy did not improve overall survival in patients OCCC cell lines (n ¼ 17). Finally, we functionally validated with OCCC. Therefore, mTORC1/2 inhibitors, currently eval- several candidate targets in OCCC cell lines and unique OCCC uated in phase II clinical trials, are proposed for the treatment patient-derived xenograft (PDX) models. Our results indicate of OCCC. Based on the mutational landscape in OCCC, our in mTORC1/2 inhibition as an approach to guide future develop- vitro results, and the toxicity observed with dual inhibitors of ment of therapeutic strategies for OCCC. PI3K and mTORC1/2, future treatment combinations of mTORC1/2 inhibitors with either PI3K or MEK inhibitors Materials and Methods could be considered to improve clinical benefit for patients Sample collection with OCCC. Primary tumor samples from 124 patients with OCCC and 47 paired control blood samples were prospectively collected from Belgium, Germany, Norway, Poland, the Netherlands, the UK, and the United States. All patients gave written informed consent for samples to be collected, and the corresponding ethical review contrast, patients with OCCC diagnosed at late stage respond boards approved the study. Tumor samples had to contain 40% poorly to standard platinum-based chemotherapy compared with tumor cells, of which 70% was OCCC, as determined by patients with late-stage HGS ovarian carcinoma (2). In recent experienced gynecologic oncology pathologists. We obtained years, genetic studies in relatively small patient groups have 17 human OCCC cell lines: TOV21G (ATCC); RMG1, RMG2, revealed the mutational landscape in OCCC. The SWI-SNF chro- OVMANA, OVTOKO, and HAC2 (JCRB Cell Bank); JHOC5 matin remodeling complex DNA-binding AT-rich interactive (RIKEN Cell Bank); OVCA429 (Cell Biolabs); OVSAYO, TUOC1, domain 1A gene (ARID1A) has been shown to be deleteriously KK, OVAS, SMOV2, and KOC7C (Dr. Hiroaki Itamochi, mutated in 40% to 57% of patients with OCCC, the highest Tottori University School of Medicine, Tottori, Japan); ES2 percentage found in any cancer (3, 4). Loss of ARID1A protein, (Dr. Els Berns, Erasmus MC, Rotterdam, the Netherlands); TAYA being a key component of the complex, may affect the expression (Dr. Yasushi Saga, Jichi Medical University, Yakushiji, of many genes (5). Activation of the PI3K/AKT/mTOR pathway, Shimotsuke-shi, Tochigi, Japan); and OV207 (Dr. Vijayalakshmi implicated in survival, protein synthesis, and proliferation, is Shridhar, Mayo Clinic). All cell lines were maintained in RPMI another major player in OCCC. PIK3CA, encoding the catalytic supplemented with 10% FCS, 100 mg/mL Penicillin/Streptomy- domain of PI3K, contains activating mutations in 30% to 40% of cin and 2 mM L-glutamine. All cell lines were tested by short patients with OCCC, whereas expression of the PI3K antagonist tandem repeat profiling and were mycoplasma free.
Recommended publications
  • AGC Kinases in Mtor Signaling, in Mike Hall and Fuyuhiko Tamanoi: the Enzymes, Vol
    Provided for non-commercial research and educational use only. Not for reproduction, distribution or commercial use. This chapter was originally published in the book, The Enzymes, Vol .27, published by Elsevier, and the attached copy is provided by Elsevier for the author's benefit and for the benefit of the author's institution, for non-commercial research and educational use including without limitation use in instruction at your institution, sending it to specific colleagues who know you, and providing a copy to your institution’s administrator. All other uses, reproduction and distribution, including without limitation commercial reprints, selling or licensing copies or access, or posting on open internet sites, your personal or institution’s website or repository, are prohibited. For exceptions, permission may be sought for such use through Elsevier's permissions site at: http://www.elsevier.com/locate/permissionusematerial From: ESTELA JACINTO, AGC Kinases in mTOR Signaling, In Mike Hall and Fuyuhiko Tamanoi: The Enzymes, Vol. 27, Burlington: Academic Press, 2010, pp.101-128. ISBN: 978-0-12-381539-2, © Copyright 2010 Elsevier Inc, Academic Press. Author's personal copy 7 AGC Kinases in mTOR Signaling ESTELA JACINTO Department of Physiology and Biophysics UMDNJ-Robert Wood Johnson Medical School, Piscataway New Jersey, USA I. Abstract The mammalian target of rapamycin (mTOR), a protein kinase with homology to lipid kinases, orchestrates cellular responses to growth and stress signals. Various extracellular and intracellular inputs to mTOR are known. mTOR processes these inputs as part of two mTOR protein com- plexes, mTORC1 or mTORC2. Surprisingly, despite the many cellular functions that are linked to mTOR, there are very few direct mTOR substrates identified to date.
    [Show full text]
  • Defining the Akt1 Interactome and Its Role in Regulating the Cell Cycle
    www.nature.com/scientificreports OPEN Defning the Akt1 interactome and its role in regulating the cell cycle Shweta Duggal1,3, Noor Jailkhani2, Mukul Kumar Midha2, Namita Agrawal3, Kanury V. S. Rao1 & Ajay Kumar1 Received: 13 July 2017 Cell growth and proliferation are two diverse processes yet always linked. Akt1, a serine/threonine Accepted: 8 January 2018 kinase, is a multi-functional protein implicated in regulation of cell growth, survival and proliferation. Published: xx xx xxxx Though it has a role in G1/S progression, the manner by which Akt1 controls cell cycle and blends cell growth with proliferation is not well explored. In this study, we characterize the Akt1 interactome as the cell cycle progresses from G0 to G1/S and G2 phase. For this, Akt1-overexpressing HEK293 cells were subjected to AP-MS. To distinguish between individual cell cycle stages, cells were cultured in the light, medium and heavy labelled SILAC media. We obtained 213 interacting partners of Akt1 from these studies. GO classifcation revealed that a signifcant number of proteins fall into functional classes related to cell growth or cell cycle processes. Of these, 32 proteins showed varying association with Akt1 in diferent cell cycle stages. Further analyses uncovered a subset of proteins showing counteracting efects so as to tune stage-specifc progression through the cycle. Thus, our study provides some novel perspectives on Akt1-mediated regulation of the cell cycle and ofers the framework for a detailed resolution of the downstream cellular mechanisms that are mediated by this kinase. Te mammalian cell cycle consists of an ordered series of events and is a highly coordinated and regulated pro- cess1.
    [Show full text]
  • Phosphatidylinositol-3-Kinase Related Kinases (Pikks) in Radiation-Induced Dna Damage
    Mil. Med. Sci. Lett. (Voj. Zdrav. Listy) 2012, vol. 81(4), p. 177-187 ISSN 0372-7025 DOI: 10.31482/mmsl.2012.025 REVIEW ARTICLE PHOSPHATIDYLINOSITOL-3-KINASE RELATED KINASES (PIKKS) IN RADIATION-INDUCED DNA DAMAGE Ales Tichy 1, Kamila Durisova 1, Eva Novotna 1, Lenka Zarybnicka 1, Jirina Vavrova 1, Jaroslav Pejchal 2, Zuzana Sinkorova 1 1 Department of Radiobiology, Faculty of Health Sciences in Hradec Králové, University of Defence in Brno, Czech Republic 2 Centrum of Advanced Studies, Faculty of Health Sciences in Hradec Králové, University of Defence in Brno, Czech Republic. Received 5 th September 2012. Revised 27 th November 2012. Published 7 th December 2012. Summary This review describes a drug target for cancer therapy, family of phosphatidylinositol-3 kinase related kinases (PIKKs), and it gives a comprehensive review of recent information. Besides general information about phosphatidylinositol-3 kinase superfamily, it characterizes a DNA-damage response pathway since it is monitored by PIKKs. Key words: PIKKs; ATM; ATR; DNA-PK; Ionising radiation; DNA-repair ABBREVIATIONS therapy and radiation play a pivotal role. Since cancer is one of the leading causes of death worldwide, it is DSB - double stand breaks, reasonable to invest time and resources in the enligh - IR - ionising radiation, tening of mechanisms, which underlie radio-resis - p53 - TP53 tumour suppressors, tance. PI - phosphatidylinositol. The aim of this review is to describe the family INTRODUCTION of phosphatidyinositol 3-kinases (PI3K) and its func - tional subgroup - phosphatidylinositol-3-kinase rela - An efficient cancer treatment means to restore ted kinases (PIKKs) and their relation to repairing of controlled tissue growth via interfering with cell sig - radiation-induced DNA damage.
    [Show full text]
  • Supplementary Information Method CLEAR-CLIP. Mouse Keratinocytes
    Supplementary Information Method CLEAR-CLIP. Mouse keratinocytes of the designated genotype were maintained in E-low calcium medium. Inducible cells were treated with 3 ug/ml final concentration doxycycline for 24 hours before performing CLEAR-CLIP. One 15cm dish of confluent cells was used per sample. Cells were washed once with cold PBS. 10mls of cold PBS was then added and cells were irradiated with 300mJ/cm2 UVC (254nM wavelength). Cells were then scraped from the plates in cold PBS and pelleted by centrifugation at 1,000g for 2 minutes. Pellets were frozen at -80oC until needed. Cells were then lysed on ice with occasional vortexing in 1ml of lysis buffer (50mM Tris-HCl pH 7.4, 100mM NaCl, 1mM MgCl2, 0.1 mM CaCl2, 1% NP-40, 0.5% Sodium Deoxycholate, 0.1% SDS) containing 1X protease inhibitors (Roche #88665) and RNaseOUT (Invitrogen #10777019) at 4ul/ml final concentration. Next, TurboDNase (Invitrogen #AM2238, 10U), RNase A (0.13ug) and RNase T1 (0.13U) were added and samples were incubated at 37oC for 5 minutes with occasional mixing. Samples were immediately placed on ice and then centrifuged at 16,160g at 4oC for 20 minutes to clear lysate. 25ul of Protein-G Dynabeads (Invitrogen #10004D) were used per IP. Dynabeads were pre-washed with lysis buffer and pre- incubated with 3ul of Wako Anti-Mouse-Ago2 (2D4) antibody. The dynabead/antibody mixture was added to the lysate and rocked for 2 hours at 4oC. All steps after the IP were done on bead until samples were loaded into the polyacrylamide gel.
    [Show full text]
  • Coordinate Phosphorylation of Multiple Residues on Single AKT1 and AKT2 Molecules
    Oncogene (2014) 33, 3463–3472 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc ORIGINAL ARTICLE Coordinate phosphorylation of multiple residues on single AKT1 and AKT2 molecules H Guo1,6, M Gao1,6,YLu1, J Liang1, PL Lorenzi2, S Bai3, DH Hawke4,JLi1, T Dogruluk5, KL Scott5, E Jonasch3, GB Mills1 and Z Ding1 Aberrant AKT activation is prevalent across multiple human cancer lineages providing an important new target for therapy. Twenty- two independent phosphorylation sites have been identified on specific AKT isoforms likely contributing to differential isoform regulation. However, the mechanisms regulating phosphorylation of individual AKT isoform molecules have not been elucidated because of the lack of robust approaches able to assess phosphorylation of multiple sites on a single AKT molecule. Using a nanofluidic proteomic immunoassay (NIA), consisting of isoelectric focusing followed by sensitive chemiluminescence detection, we demonstrate that under basal and ligand-induced conditions that the pattern of phosphorylation events is markedly different between AKT1 and AKT2. Indeed, there are at least 12 AKT1 peaks and at least 5 AKT2 peaks consistent with complex combinations of phosphorylation of different sites on individual AKT molecules. Following insulin stimulation, AKT1 was phosphorylated at Thr308 in the T-loop and Ser473 in the hydrophobic domain. In contrast, AKT2 was only phosphorylated at the equivalent sites (Thr309 and Ser474) at low levels. Further, Thr308 and Ser473 phosphorylation occurred predominantly on the same AKT1 molecules, whereas Thr309 and Ser474 were phosphorylated primarily on different AKT2 molecules. Although basal AKT2 phosphorylation was sensitive to inhibition of phosphatidylinositol 3-kinase (PI3K), basal AKT1 phosphorylation was essentially resistant.
    [Show full text]
  • The PRKCI and SOX2 Oncogenes Are Coamplified and Cooperate to Activate Hedgehog Signaling in Lung Squamous Cell Carcinoma
    Cancer Cell Article The PRKCI and SOX2 Oncogenes Are Coamplified and Cooperate to Activate Hedgehog Signaling in Lung Squamous Cell Carcinoma Verline Justilien,1 Michael P. Walsh,1 Syed A. Ali,1 E. Aubrey Thompson,1 Nicole R. Murray,1 and Alan P. Fields1,* 1Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL 32224, USA *Correspondence: fi[email protected] http://dx.doi.org/10.1016/j.ccr.2014.01.008 SUMMARY We report that two oncogenes coamplified on chromosome 3q26, PRKCI and SOX2, cooperate to drive a stem-like phenotype in lung squamous cell carcinoma (LSCC). Protein kinase Ci (PKCi) phosphorylates SOX2, a master transcriptional regulator of stemness, and recruits it to the promoter of Hedgehog (Hh) acyltransferase (HHAT) that catalyzes the rate-limiting step in Hh ligand production. PKCi-mediated SOX2 phosphorylation is required for HHAT promoter occupancy, HHAT expression, and maintenance of a stem-like phenotype. Primary LSCC tumors coordinately overexpress PKCi, SOX2, and HHAT and require PKCi-SOX2-HHAT signaling to maintain a stem-like phenotype. Thus, PKCi and SOX2 are genetically, biochemically, and functionally linked in LSCC, and together they drive tumorigenesis by establishing a cell-autonomous Hh signaling axis. INTRODUCTION Similar cell populations exist in several cancer types (Chen et al., 2012; Driessens et al., 2012; Schepers et al., 2012). Line- Lung cancer is the major cause of cancer death, with a 5-year- age tracing reveals these cells clonally expand to give rise to survival rate of 16% (Siegel et al., 2012). Non-small cell lung malignant and nonmalignant, differentiated cell types.
    [Show full text]
  • Expressed Gene Fusions As Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer
    Published OnlineFirst December 14, 2017; DOI: 10.1158/2159-8290.CD-17-0535 RESEARCH ARTICLE Expressed Gene Fusions as Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer Karina J. Matissek1,2, Maristela L. Onozato3, Sheng Sun1,2, Zongli Zheng2,3,4, Andrew Schultz1, Jesse Lee3, Kristofer Patel1, Piiha-Lotta Jerevall2,3, Srinivas Vinod Saladi1,2, Allison Macleay3, Mehrad Tavallai1,2, Tanja Badovinac-Crnjevic5, Carlos Barrios6, Nuran Beşe7, Arlene Chan8, Yanin Chavarri-Guerra9, Marcio Debiasi6, Elif Demirdögen10, Ünal Egeli10, Sahsuvar Gökgöz10, Henry Gomez11, Pedro Liedke6, Ismet Tasdelen10, Sahsine Tolunay10, Gustavo Werutsky6, Jessica St. Louis1, Nora Horick12, Dianne M. Finkelstein2,12, Long Phi Le2,3, Aditya Bardia1,2, Paul E. Goss1,2, Dennis C. Sgroi2,3, A. John Iafrate2,3, and Leif W. Ellisen1,2 ABSTRACT We sought to uncover genetic drivers of hormone receptor–positive (HR+) breast cancer, using a targeted next-generation sequencing approach for detecting expressed gene rearrangements without prior knowledge of the fusion partners. We identified inter- genic fusions involving driver genes, including PIK3CA, AKT3, RAF1, and ESR1, in 14% (24/173) of unselected patients with advanced HR+ breast cancer. FISH confirmed the corresponding chromo- somal rearrangements in both primary and metastatic tumors. Expression of novel kinase fusions in nontransformed cells deregulates phosphoprotein signaling, cell proliferation, and survival in three- dimensional culture, whereas expression in HR+ breast cancer models modulates estrogen-dependent growth and confers hormonal therapy resistance in vitro and in vivo. Strikingly, shorter overall survival was observed in patients with rearrangement-positive versus rearrangement-negative tumors. Cor- respondingly, fusions were uncommon (<5%) among 300 patients presenting with primary HR+ breast cancer.
    [Show full text]
  • Diverse Mechanisms Activate the PI 3-Kinase/Mtor Pathway In
    Tran et al. BMC Cancer (2021) 21:136 https://doi.org/10.1186/s12885-021-07826-4 RESEARCH ARTICLE Open Access Diverse mechanisms activate the PI 3- kinase/mTOR pathway in melanomas: implications for the use of PI 3-kinase inhibitors to overcome resistance to inhibitors of BRAF and MEK Khanh B. Tran1,2,3, Sharada Kolekar1, Anower Jabed2, Patrick Jaynes2, Jen-Hsing Shih2, Qian Wang2, Jack U. Flanagan1,3, Gordon W. Rewcastle1,3, Bruce C. Baguley1,3 and Peter R. Shepherd1,2,3* Abstract Background: The PI 3-kinase (PI3K) pathway has been implicated as a target for melanoma therapy. Methods: Given the high degree of genetic heterogeneity in melanoma, we sought to understand the breadth of variation in PI3K signalling in the large NZM panel of early passage cell lines developed from metastatic melanomas. Results: We find the vast majority of lines show upregulation of this pathway, and this upregulation is achieved by a wide range of mechanisms. Expression of all class-IA PI3K isoforms was readily detected in these cell lines. A range of genetic changes in different components of the PI3K pathway was seen in different lines. Coding variants or amplification were identified in the PIK3CA gene, and amplification of the PK3CG gene was common. Deletions in the PIK3R1 and PIK3R2 regulatory subunits were also relatively common. Notably, no genetic variants were seen in the PIK3CD gene despite p110δ being expressed in many of the lines. Genetic variants were detected in a number of genes that encode phosphatases regulating the PI3K signalling, with reductions in copy number common in PTEN, INPP4B, INPP5J, PHLLP1 and PHLLP2 genes.
    [Show full text]
  • Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase
    cells Review Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase Jaroslav Kalous *, Denisa Jansová and Andrej Šušor Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic; [email protected] (D.J.); [email protected] (A.Š.) * Correspondence: [email protected] Received: 28 April 2020; Accepted: 24 June 2020; Published: 27 June 2020 Abstract: Cyclin dependent kinase 1 (CDK1) has been primarily identified as a key cell cycle regulator in both mitosis and meiosis. Recently, an extramitotic function of CDK1 emerged when evidence was found that CDK1 is involved in many cellular events that are essential for cell proliferation and survival. In this review we summarize the involvement of CDK1 in the initiation and elongation steps of protein synthesis in the cell. During its activation, CDK1 influences the initiation of protein synthesis, promotes the activity of specific translational initiation factors and affects the functioning of a subset of elongation factors. Our review provides insights into gene expression regulation during the transcriptionally silent M-phase and describes quantitative and qualitative translational changes based on the extramitotic role of the cell cycle master regulator CDK1 to optimize temporal synthesis of proteins to sustain the division-related processes: mitosis and cytokinesis. Keywords: CDK1; 4E-BP1; mTOR; mRNA; translation; M-phase 1. Introduction 1.1. Cyclin Dependent Kinase 1 (CDK1) Is a Subunit of the M Phase-Promoting Factor (MPF) CDK1, a serine/threonine kinase, is a catalytic subunit of the M phase-promoting factor (MPF) complex which is essential for cell cycle control during the G1-S and G2-M phase transitions of eukaryotic cells.
    [Show full text]
  • Role of Ataxia-Telangiectasia Mutated Kinase in Cardiac Autophagy and Glucose Metabolism Under Ischemic Conditions Patsy Thrasher East Tennessee State University
    East Tennessee State University Digital Commons @ East Tennessee State University Electronic Theses and Dissertations Student Works 8-2018 Role of Ataxia-Telangiectasia Mutated Kinase in Cardiac Autophagy and Glucose Metabolism Under Ischemic Conditions Patsy Thrasher East Tennessee State University Follow this and additional works at: https://dc.etsu.edu/etd Part of the Biology Commons, and the Physiology Commons Recommended Citation Thrasher, Patsy, "Role of Ataxia-Telangiectasia Mutated Kinase in Cardiac Autophagy and Glucose Metabolism Under Ischemic Conditions" (2018). Electronic Theses and Dissertations. Paper 3442. https://dc.etsu.edu/etd/3442 This Dissertation - Open Access is brought to you for free and open access by the Student Works at Digital Commons @ East Tennessee State University. It has been accepted for inclusion in Electronic Theses and Dissertations by an authorized administrator of Digital Commons @ East Tennessee State University. For more information, please contact [email protected]. Role of Ataxia-Telangiectasia Mutated Kinase in Cardiac Autophagy and Glucose Metabolism Under Ischemic Conditions ____________________________________ A dissertation presented to the faculty of the Department of Biomedical Sciences East Tennessee State University In partial fulfillment of the requirements for the degree Doctor of Philosophy in Biomedical Sciences __________________________________ by Patsy R. Thrasher August 2018 _________________________________ Krishna Singh, Ph.D., Chair Mahipal Singh, Ph.D. Chuanfu Li, M.D. Tom Ecay, Ph.D. Yue Zou, Ph.D. Douglas Thewke, Ph.D. Keywords: ATM, myocardial infarction, autophagy, ischemia, glucose, metabolism ABSTRACT Role of Ataxia-Telangiectasia Mutated Kinase in Cardiac Autophagy and Glucose Metabolism Under Ischemic Conditions by Patsy R. Thrasher Ataxia-telangiectasia mutated kinase (ATM), a serine/threonine kinase primarily located in the nucleus, is typically activated in response to DNA damage.
    [Show full text]
  • Analysis of 3-Phosphoinositide-Dependent Kinase-1 Signaling and Function in ES Cells
    EXPERIMENTAL CELL RESEARCH 314 (2008) 2299– 2312 available at www.sciencedirect.com www.elsevier.com/locate/yexcr Research Article Analysis of 3-phosphoinositide-dependent kinase-1 signaling and function in ES cells Tanja Tamgüneya,b, Chao Zhangc, Dorothea Fiedlerc, Kevan Shokatc, David Stokoea,⁎ aUCSF Cancer Research Institute, USA bMolecular Medicine Program, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany cDepartment of Cellular and Molecular Pharmacology, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94115, USA ARTICLE INFORMATION ABSTRACT Article Chronology: 3-Phosphoinositide-dependent kinase-1 (PDK1) phosphorylates and activates several Received 5 February 2008 kinases in the cAMP-dependent, cGMP-dependent and protein kinase C (AGC) family. Revised version received Many putative PDK1 substrates have been identified, but have not been analyzed following 15 April 2008 transient and specific inhibition of PDK1 activity. Here, we demonstrate that a previously Accepted 16 April 2008 characterized PDK1 inhibitor, BX-795, shows biological effects that are not consistent with Available online 23 April 2008 PDK1 inhibition. Therefore, we describe the creation and characterization of a PDK1 mutant, L159G, which can bind inhibitor analogues containing bulky groups that hinder access to the − − Keywords: ATP binding pocket of wild type (WT) kinases. When expressed in PDK1 / ES cells, PDK1 PDK1 L159G restored phosphorylation of PDK1 targets known to be hypophosphorylated in these PKB/Akt cells. Screening of multiple inhibitor analogues showed that 1-NM-PP1 and 3,4-DMB-PP1 − − PI3K optimally inhibited the phosphorylation of PDK1 targets in PDK1 / ES cells expressing PDK1 Chemical genetics L159G but not WT PDK1. These compounds confirmed previously assumed PDK1 substrates, AGC kinases but revealed distinct dephosphorylation kinetics.
    [Show full text]
  • CDK5 Functions As a Tumor Promoter in Human Lung Cancer Jie Zeng1#, Shuanshuan Xie1#, Yang Liu1, Changxing Shen1, Xiaolian Song1, Guo-Lei Zhou2, 3, Changhui Wang1
    Journal of Cancer 2018, Vol. 9 3950 Ivyspring International Publisher Journal of Cancer 2018; 9(21): 3950-3961. doi: 10.7150/jca.25967 Research Paper CDK5 Functions as a Tumor Promoter in Human Lung Cancer Jie Zeng1#, Shuanshuan Xie1#, Yang Liu1, Changxing Shen1, Xiaolian Song1, Guo-Lei Zhou2, 3, Changhui Wang1 1. Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, PR China; 2. Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA; 3. Molecular Biosciences Program, Arkansas State University, State University, AR 72467, USA. # These authors have contributed equally to this work. Corresponding author: Changhui Wang, No.301, Mid Yanchang Rd, Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China, 200072. Email: [email protected], Fax number: 86-021-66301685, Telephone: 86-021-66301685 © Ivyspring International Publisher. This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions. Received: 2018.03.09; Accepted: 2018.08.19; Published: 2018.10.10 Abstract Cyclin-dependent kinase 5 (CDK5), an atypical member of the cyclin-dependent kinase family, plays an important role in the nervous system. Recent studies have shown that CDK5 is also associated with tumors. However, few studies have been done to investigate the mechanism underlying the connection between CDK5 and cancers. To explore the role of CDK5 in cancers by using an extensive bioinformatics data mining process. We mined the transcriptional, survival, functions and structure of CDK5 gene through databases and in vitro experiments.
    [Show full text]